Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
Add more filters

Publication year range
1.
Soft Matter ; 10(12): 2059-68, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24800271

ABSTRACT

The assembly kinetics of intermediate filament (IF) proteins from tetrameric complexes to single filaments and networks depends on the protein concentration, temperature and the ionic composition of their environment. We systematically investigate how changes in the concentration of monovalent potassium and divalent magnesium ions affect the internal organization of the resulting filaments. Small angle X-ray scattering (SAXS) is very sensitive to changes in the filament cross-section such as diameter or compactness. Our measurements reveal that filaments formed in the presence of magnesium chloride differ distinctly from filaments formed in the presence of potassium chloride. The principle multi-step assembly mechanism from tetramers via unit-length filaments (ULF) to elongated filaments is not changed by the valency of ions. However, the observed differences indicate that the magnesium ions free the head domains of tetramers from unproductive interactions to allow assembly but at the same time mediate strong inter-tetrameric interactions that impede longitudinal annealing of unit-length filaments considerably, thus slowing down filament growth.


Subject(s)
Intermediate Filament Proteins/chemistry , Intermediate Filaments/ultrastructure , Scattering, Small Angle , Vimentin/chemistry , Cytoskeleton/chemistry , Cytoskeleton/ultrastructure , Intermediate Filament Proteins/ultrastructure , Intermediate Filaments/chemistry , Ions/chemistry , Kinetics , Vimentin/ultrastructure , X-Ray Diffraction
2.
Nat Struct Mol Biol ; 31(6): 939-949, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38632361

ABSTRACT

Intermediate filaments (IFs) are integral components of the cytoskeleton. They provide cells with tissue-specific mechanical properties and are involved in numerous cellular processes. Due to their intricate architecture, a 3D structure of IFs has remained elusive. Here we use cryo-focused ion-beam milling, cryo-electron microscopy and tomography to obtain a 3D structure of vimentin IFs (VIFs). VIFs assemble into a modular, intertwined and flexible helical structure of 40 α-helices in cross-section, organized into five protofibrils. Surprisingly, the intrinsically disordered head domains form a fiber in the lumen of VIFs, while the intrinsically disordered tails form lateral connections between the protofibrils. Our findings demonstrate how protein domains of low sequence complexity can complement well-folded protein domains to construct a biopolymer with striking mechanical strength and stretchability.


Subject(s)
Cryoelectron Microscopy , Intermediate Filaments , Vimentin , Vimentin/chemistry , Vimentin/metabolism , Vimentin/ultrastructure , Intermediate Filaments/chemistry , Intermediate Filaments/metabolism , Intermediate Filaments/ultrastructure , Humans , Models, Molecular , Protein Domains , Protein Conformation, alpha-Helical
3.
J Struct Biol ; 177(1): 46-53, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22119849

ABSTRACT

Cytoskeletal intermediate filaments (IFs) assemble from the elementary dimers based on a segmented α-helical coiled-coil (CC) structure. Crystallographic studies of IF protein fragments remain the main route to access their atomic structure. To enable crystallization, such fragments must be sufficiently short. As a consequence, they often fail to assemble into the correct CC dimers. In particular, human vimentin fragment D3 corresponding to the first half of coil2 (residues 261-335) stays monomeric in solution. We have induced its dimerization via introducing a disulfide link between two cysteines engineered in the hydrophobic core of the CC close to its N-terminus. The 2.3 Å crystal structure of the D3st (stabilized) fragment reveals a mostly parallel α-helical bundle structure in its N-terminal half which smoothly continues into a left-handed CC towards the C-terminus. This provides a direct evidence for a continuously α-helical structure of the coil2 segment and disproves the previously suggested existence of linker L2 separating it into two left-handed CCs. The general principles of CC dimer stabilization by disulfide introduction are also discussed.


Subject(s)
Disulfides/chemistry , Vimentin/metabolism , Vimentin/ultrastructure , Amino Acid Sequence , Crystallization , Crystallography, X-Ray , Disulfides/analysis , Humans , Models, Molecular , Molecular Sequence Data , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary , Vimentin/analysis
4.
J Struct Biol ; 177(1): 14-23, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22120848

ABSTRACT

Although intermediate filaments are one of three major cytoskeletal systems of vertebrate cells, they remain the least understood with respect to their structure and function. This is due in part to the fact that they are encoded by a large gene family which is developmentally regulated in a cell and tissue type specific fashion. This article is in honor of Ueli Aebi. It highlights the studies on IF that have been carried out by our laboratory for more than 40 years. Many of our advances in understanding IF are based on conversations with Ueli which have taken place during adventurous and sometimes dangerous hiking and biking trips throughout the world.


Subject(s)
Intermediate Filaments/metabolism , Intermediate Filaments/ultrastructure , Animals , Cell Movement , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Keratins/metabolism , Keratins/ultrastructure , Phosphorylation , Vimentin/metabolism , Vimentin/ultrastructure
5.
J Biomed Biotechnol ; 2011: 528276, 2011.
Article in English | MEDLINE | ID: mdl-22131810

ABSTRACT

Serum amyloid A (SAA) acts as a major acute phase protein and represents a sensitive and accurate marker of inflammation. Besides its hepatic origin, as the main source of serum SAA, this protein is also produced extrahepatically. The mRNA levels of SAA become significantly elevated following proinflammatory stimuli, as well as, are induced through their own positive feedback in human primary coronary artery endothelial cells. However, the intracellular functions of SAA are so far unknown. Colocalization of SAA with cytoskeletal filaments has previously been proposed, so we analyzed the colocalization of SAA with all three cytoskeletal elements: actin filaments, vimentin filaments, and microtubules. Immunofluorescent double-labeling analyses confirmed by PLA method revealed a strict colocalization of SAA with microtubules and a very infrequent attachment to vimentin while the distribution of actin filaments appeared clearly separated from SAA staining. Also, no significant colocalization was found between SAA and endomembranes labeled with the fluorescent lipid stain DiO6. However, SAA appears to be located also unbound in the cytosol, as well as inside the nucleus and within nanotubes extending from the cells or bridging neighboring cells. These different locations of SAA in endothelial cells strongly indicate multiple potential functions of this protein.


Subject(s)
Actin Cytoskeleton/metabolism , Coronary Vessels/metabolism , Endothelium, Vascular/metabolism , Microtubules/metabolism , Serum Amyloid A Protein/metabolism , Vimentin/metabolism , Actin Cytoskeleton/diagnostic imaging , Cell Nucleus/ultrastructure , Coronary Vessels/cytology , Cytosol/ultrastructure , Fluorescent Antibody Technique, Indirect , Humans , Inflammation/metabolism , Microtubules/ultrastructure , Serum Amyloid A Protein/chemistry , Staining and Labeling , Ultrasonography , Vimentin/ultrastructure
6.
J Struct Biol ; 170(2): 369-76, 2010 May.
Article in English | MEDLINE | ID: mdl-20176112

ABSTRACT

Intermediate filaments (IFs) are essential cytoskeletal components in metazoan cells. They assemble from elementary dimers that are built around the central alpha-helical coiled-coil rod domain representing the IF 'signature'. The rod consists of two similarly-sized parts, coil 1 and coil 2, connected by a non-alpha-helical linker L12. Coil 2 is absolutely conserved in length across all IF types and was initially predicted to consist of a short coiled-coil segment 2A based on a heptad pattern of hydrophobic residues, another linker L2 and a coiled-coil segment 2B. Here we present the crystal structure of human vimentin fragment including residues 261-335 i.e. approximately the first half of coil 2. The N-terminal part of this fragment reveals a parallel alpha-helical bundle characterized by 3.5 consecutive hendecad repeats. It is immediately followed by a regular left-handed coiled coil. The distinct non-helical linker L2 is therefore not observed. Together with the previously determined crystal structure of the major part of segment 2B (Strelkov et al., 2002), we can now build a complete atomic model of the 21nm long vimentin coil 2 dimer being a relatively rigid rod.


Subject(s)
Protein Structure, Secondary , Vimentin/ultrastructure , Amino Acid Sequence , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Sequence Data , Molecular Structure , Peptide Fragments/chemistry , Peptide Fragments/genetics , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Tertiary , Sequence Alignment
7.
Cell Motil Cytoskeleton ; 66(11): 986-99, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19530175

ABSTRACT

Short polypeptides from intermediate filament (IF) proteins containing one of the two IF-consensus motifs interfere severely with filament assembly in vitro. We now have systematically investigated a series of larger fragments of the muscle-specific IF protein desmin representing entire functional domains such as coil1 or coil 2. "Half molecules" comprising the amino-terminal portion of desmin, such as DesDeltaC240 and the "tagged" derivative Des(ESA)DeltaC244, assembled into large, roundish aggregates already at low ionic strength, DesDeltaC250 formed extended, relatively uniform filaments, whereas DesDeltaC265 and DesDeltaC300 were soluble under these conditions. Surprisingly, all mutant desmin fragments assembled very rapidly into long thick filaments or spacious aggregates when the ionic strength was raised to standard assembly conditions. In contrast, when these desmin mutants were assembled in the presence of wild-type (WT) desmin, their assembly properties were completely changed: The elongation of the two shorter desmin fragments was completely inhibited by WT desmin, whereas DesDeltaC250, DesDeltaC265 and DesDeltaC300 coassembled with desmin into filaments, but these mixed filaments were distinctly disturbed and exhibited a very different phenotype for each mutant. After transfection into fibroblasts and cardiomyocytes, the truncated mutant Des (ESA)DeltaC244 localized largely to the cytoplasm, as revealed by a tag-specific monoclonal antibody, and also partially colocalized there with the collapsed endogenous vimentin and desmin systems indicating its interference with IF-organizing processes. In contrast, in cells without an authentic cytoplasmic IF system such as line SW13, Des(ESA)DeltaC242 entered the nucleus and was deposited in small dot-like structures in chromatin-free spaces without any noticeable effect on nuclear morphology.


Subject(s)
Desmin/chemistry , Desmin/metabolism , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutant Proteins/ultrastructure , Protein Interaction Domains and Motifs/physiology , 3T3 Cells , Adrenocortical Carcinoma/metabolism , Adrenocortical Carcinoma/ultrastructure , Animals , Cell Line, Transformed , Desmin/ultrastructure , Heart Atria/metabolism , Heart Atria/ultrastructure , Humans , Mice , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Protein Multimerization , Structure-Activity Relationship , Vimentin/metabolism , Vimentin/ultrastructure
8.
Mol Neurobiol ; 57(3): 1484-1501, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31773411

ABSTRACT

The leptomeninges, referring to the arachnoid and pia mater and their projections into the perivascular compartments in the central nervous system, actively participate in diverse biological processes including fluid homeostasis, immune cell infiltrations, and neurogenesis, yet their detailed cellular and molecular identities remain elusive. This study aimed to characterize platelet-derived growth factor beta (PDGFR-ß)-expressing cells in the leptomeninges in the adult rat brain using light and electron microscopy. PDGFR-ß+ cells were observed in the inner arachnoid, arachnoid trabeculae, pia mater, and leptomeningeal sheath of the subarachnoid vessels, thereby forming a cellular network throughout the leptomeninges. Leptomeningeal PDGFR-ß+ cells were commonly characterized by large euchromatic nuclei, thin branching processes forming web-like network, and the expression of the intermediate filaments nestin and vimentin. These cells were typical of active fibroblasts with a well-developed rough endoplasmic reticulum and close spatial correlation with collagen fibrils. Leptomeningeal PDGFR-ß+ cells ensheathing the vasculature in the subarachnoid space joined with pial PDGFR-ß+ cells upon entering the cortical parenchyma, yet perivascular PDGFR-ß+ cells in these penetrating vessels underwent abrupt changes in their morphological and molecular characteristics: they became more flattened with loss of immunoreactivity for nestin and vimentin and deficient collagen deposition, which was indicative of inactive fibroblasts termed fibrocytes. In the cortical parenchyma, PDGFR-ß immunoreactivity was almost exclusively localized to larger caliber vessels, and significantly decreased in capillary-like microvessels. Collectively, our data identify PDGFR-ß as a novel cellular marker for leptomeningeal fibroblasts comprising the leptomeninges and perivascular adventitial cells of the subarachnoid and penetrating large-sized cortical vasculatures.


Subject(s)
Arachnoid/metabolism , Brain/ultrastructure , Meninges/metabolism , Meninges/ultrastructure , Animals , Arachnoid/ultrastructure , Brain/metabolism , Collagen/metabolism , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Microscopy, Electron/methods , Pia Mater/pathology , Pia Mater/ultrastructure , Proto-Oncogene Proteins c-sis/metabolism , Rats , Vimentin/metabolism , Vimentin/ultrastructure
9.
J Cell Biol ; 113(5): 1111-24, 1991 Jun.
Article in English | MEDLINE | ID: mdl-1710225

ABSTRACT

Vimentin and keratin are coexpressed in many cells, but they segregate into two distinct intermediate filament (IF) networks. To understand the molecular basis for the sorting out of these IF subunits, we genetically engineered cDNAs encoding hybrid IF proteins composed of part vimentin and part type I keratin. When these cDNAs were transiently expressed in cells containing vimentin, keratin, or both IFs, the hybrid IF proteins all recognized one or the other or both networks. The ability to distinguish networks was dependent upon which segments of IF proteins were present in each construct. Constructs containing sequences encoding either helix 1B or helix 2B seemed to be the most critical in conferring IF recognition. At least for type I keratins, recognition was exerted at the level of dimer formation with wild-type type II keratin, as demonstrated by anion exchange chromatography. Interestingly, despite the fact that swapping of helical domains was not as deleterious to IF structure/function as deletion of helical domains, keratin/vimentin hybrids still caused structural aberrations in one or more of the cytoplasmic IF network. Thus, sequence diversity among IF proteins seems to influence not only coiled-coil but also higher ordered associations leading to 10-nm filament formation and/or IF interactions with other cellular organelles/proteins.


Subject(s)
Intermediate Filaments/physiology , Keratins/physiology , Vimentin/physiology , Amino Acid Sequence , Animals , Cell Line , DNA/genetics , Humans , Intermediate Filaments/ultrastructure , Keratins/genetics , Keratins/ultrastructure , Molecular Sequence Data , Protein Multimerization , Sequence Homology, Nucleic Acid , Transfection , Vimentin/genetics , Vimentin/ultrastructure
10.
J Cell Biol ; 134(6): 1455-67, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8830774

ABSTRACT

We have generated a series of plectin deletion and mutagenized cDNA constructs to dissect the functional sequences that mediate plectin's interaction with intermediate filament (IF) networks, and scored their ability to coalign or disrupt intermediate filaments when ectopically expressed in rat kangaroo PtK2 cells. We show that a stretch of approximately 50 amino acid residues within plectin's carboxy-terminal repeat 5 domain serves as a unique binding site for both vimentin and cytokeratin IF networks of PtK2 cells. Part of the IF-binding domain was found to constitute a functional nuclear localization signal (NLS) motif, as demonstrated by nuclear import of cytoplasmic proteins linked to this sequence. Site directed mutagenesis revealed a specific cluster of four basic amino acid residues (arg4277-arg4280) residing within the NLS sequence motif to be essential for IF binding. When mutant proteins corresponding to those expressed in PtK2 cells were expressed in bacteria and purified proteins subjected to a sensitive quantitative overlay binding assay using Eu3+-labeled vimentin, the relative binding capacities of mutant proteins measured were fully consistent with the mutant's phenotypes observed in living cells. Using recombinant proteins we also show by negative staining and rotary shadowing electron microscopy that in vitro assembled vimentin intermediate filaments become packed into dense aggregates upon incubation with plectin repeat 5 domain, in contrast to repeat 4 domain or a mutated repeat 5 domain.


Subject(s)
Amino Acids/chemistry , Cell Nucleus/metabolism , Intermediate Filament Proteins/chemistry , Vimentin/chemistry , Amino Acid Sequence , Animals , Bacterial Physiological Phenomena , Base Sequence , Binding Sites/physiology , Biological Transport/physiology , Cell Line/physiology , Cytoplasm/chemistry , Cytoplasm/physiology , Cytoskeleton/chemistry , Gene Expression/physiology , Intermediate Filament Proteins/metabolism , Intermediate Filament Proteins/ultrastructure , Keratinocytes/physiology , Keratins/metabolism , Macropodidae , Mice , Microscopy, Electron , Molecular Sequence Data , Mutagenesis, Site-Directed , Plectin , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Vimentin/metabolism , Vimentin/ultrastructure
11.
J Cell Biol ; 126(2): 445-56, 1994 Jul.
Article in English | MEDLINE | ID: mdl-7518466

ABSTRACT

In most myogenic systems, synthesis of the intermediate filament (IF) protein vimentin precedes the synthesis of the muscle-specific IF protein desmin. In the dorsal myotome of the Xenopus embryo, however, there is no preexisting vimentin filament system and desmin's initial organization is quite different from that seen in vimentin-containing myocytes (Cary and Klymkowsky, 1994. Differentiation. In press.). To determine whether the organization of IFs in the Xenopus myotome reflects features unique to Xenopus or is due to specific properties of desmin, we used the injection of plasmid DNA to drive the synthesis of vimentin or desmin in myotomal cells. At low levels of accumulation, exogenous vimentin and desmin both enter into the endogenous desmin system of the myotomal cell. At higher levels exogenous vimentin forms longitudinal IF systems similar to those seen in vimentin-expressing myogenic systems and massive IF bundles. Exogenous desmin, on the other hand, formed a reticular IF meshwork and non-filamentous aggregates. In embryonic epithelial cells, both vimentin and desmin formed extended IF networks. Vimentin and desmin differ most dramatically in their NH2-terminal "head" regions. To determine whether the head region was responsible for the differences in the behavior of these two proteins, we constructed plasmids encoding chimeric proteins in which the head of one was attached to the body of the other. In muscle, the vimentin head-desmin body (VDD) polypeptide formed longitudinal IFs and massive IF bundles like vimentin. The desmin head-vimentin body (DVV) polypeptide, on the other hand, formed IF meshworks and non-filamentous structures like desmin. In embryonic epithelial cells DVV formed a discrete filament network while VDD did not. Based on the behavior of these chimeric proteins, we conclude that the head domains of vimentin and desmin are structurally distinct and not interchangeable, and that the head domain of desmin is largely responsible for desmin's muscle-specific behaviors.


Subject(s)
Desmin/ultrastructure , Intermediate Filaments/ultrastructure , Muscles/chemistry , Vimentin/ultrastructure , Amino Acid Sequence , Animals , Antibodies, Monoclonal , Base Sequence , Cells, Cultured , DNA, Recombinant , Desmin/analysis , Desmin/chemistry , Desmin/genetics , Epitopes/analysis , Humans , Microinjections , Molecular Sequence Data , Muscles/cytology , Protein Structure, Tertiary , Proto-Oncogene Proteins c-myc , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/ultrastructure , Vimentin/analysis , Vimentin/chemistry , Vimentin/genetics , Xenopus/embryology , Xenopus/genetics , Zygote/chemistry
12.
J Mol Biol ; 360(3): 623-30, 2006 Jul 14.
Article in English | MEDLINE | ID: mdl-16765985

ABSTRACT

Intermediate filaments (IFs), together with actin filaments and microtubules, compose the cytoskeleton. Among other functions, IFs impart mechanical stability to cells when exposed to mechanical stress and act as a support when the other cytoskeletal filaments cannot keep the structural integrity of the cells. Here we present a study on the bending properties of single vimentin IFs in which we used an atomic force microscopy (AFM) tip to elastically deform single filaments hanging over a porous membrane. We obtained a value for the bending modulus of non-stabilized IFs between 300 MPa and 400 MPa. Our results together with previous ones suggest that IFs present axial sliding between their constitutive building blocks and therefore have a bending modulus that depends on the filament length. Measurements of glutaraldehyde-stabilized filaments were also performed to reduce the axial sliding between subunits and therefore provide a lower limit estimate of the Young's modulus of the filaments. The results show an increment of two to three times in the bending modulus for the stabilized IFs with respect to the non-stabilized ones, suggesting that the Young's modulus of vimentin IFs should be around 900 MPa or higher.


Subject(s)
Intermediate Filaments/chemistry , Intermediate Filaments/ultrastructure , Vimentin/chemistry , Vimentin/ultrastructure , Aluminum Oxide/chemistry , Animals , Biomechanical Phenomena , Cricetinae , Microscopy, Atomic Force , Thermodynamics
13.
Mol Biol Cell ; 10(6): 1997-2015, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10359611

ABSTRACT

The intermediate filament protein vimentin is a major phosphoprotein in mammalian fibroblasts, and reversible phosphorylation plays a key role in its dynamic rearrangement. Selective inhibition of type 2A but not type 1 protein phosphatases led to hyperphosphorylation and concomitant disassembly of vimentin, characterized by a collapse into bundles around the nucleus. We have analyzed the potential role of one of the major protein phosphatase 2A (PP2A) regulatory subunits, B55, in vimentin dephosphorylation. In mammalian fibroblasts, B55 protein was distributed ubiquitously throughout the cytoplasm with a fraction associated to vimentin. Specific depletion of B55 in living cells by antisense B55 RNA was accompanied by disassembly and increased phosphorylation of vimentin, as when type 2A phosphatases were inhibited using okadaic acid. The presence of B55 was a prerequisite for PP2A to efficiently dephosphorylate vimentin in vitro or to induce filament reassembly in situ. Both biochemical fractionation and immunofluorescence analysis of detergent-extracted cells revealed that fractions of PP2Ac, PR65, and B55 were tightly associated with vimentin. Furthermore, vimentin-associated PP2A catalytic subunit was displaced in B55-depleted cells. Taken together these data show that, in mammalian fibroblasts, the intermediate filament protein vimentin is dephosphorylated by PP2A, an event targeted by B55.


Subject(s)
Intermediate Filaments/metabolism , Phosphoprotein Phosphatases/metabolism , Pyrans , Spiro Compounds , Vimentin/metabolism , Antifungal Agents/pharmacology , Cytoplasm/metabolism , Enzyme Inhibitors/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Intermediate Filaments/ultrastructure , Interphase/physiology , Okadaic Acid/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/genetics , Phosphorylation , Protein Phosphatase 2 , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Vimentin/genetics , Vimentin/ultrastructure
14.
Eur J Cell Biol ; 85(1): 1-10, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16373170

ABSTRACT

To get new insights into the function of the intermediate filament (IF) protein vimentin in cell physiology, we generated two mutant cDNAs, one with a point mutation in the consensus motif in coil1A (R113C) and one with the complete deletion of coil 2B of the rod domain. In keratins and glia filament protein (GFAP), analogous mutations cause keratinopathies and Alexander disease, respectively. Both mutants prevented filament assembly in vitro and inhibited assembly of wild-type vimentin when present in equal amounts. In stably transfected preadipocytes, these mutants caused the complete disruption of the endogenous vimentin network, demonstrating their dominant-negative behaviour. Cytoplasmic vimentin aggregates colocalised with the chaperones alphaB-crystallin and HSP40. Moreover, vimR113C mutant cells were more resistant against staurosporine-induced apoptosis compared to controls. We hypothesise that mutations in the vimentin gene, like in most classes of IF genes, may contribute to distinct human diseases.


Subject(s)
Apoptosis , Cytoskeleton/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Mutation/genetics , Vimentin/genetics , Vimentin/metabolism , 3T3 Cells , Animals , Cells, Cultured , Cytoplasm , Inclusion Bodies/metabolism , Mice , Molecular Chaperones/metabolism , Multiprotein Complexes , Protein Transport , Subcellular Fractions , Vimentin/ultrastructure , Viscosity
15.
Acta Otolaryngol ; 126(8): 801-10, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16846921

ABSTRACT

CONCLUSIONS: These experimental findings suggest the feasibility of artificial middle ear mucosa grafting as an effective treatment for achieving mucosal regeneration after middle ear surgery. OBJECTIVES: Postoperative mucosal regeneration of tympanic cavity and mastoid cavity is of great importance after middle ear surgery. We reconstructed in vitro a three-dimensional middle ear mucosal organ, and assessed its feasibility for regenerative medicine of middle ear-related diseases. MATERIALS AND METHODS: Epithelial cells and fibroblasts were isolated from the middle ear mucosa of rats and propagated by subculturing. An artificial middle ear mucosal organ was reconstructed by overlaying the middle ear epithelial cells on three-dimensional lattices of a collagen gel that had been repopulated with the fibroblasts. In addition, the artificial organ was implanted in the middle ear cavity of rats. RESULTS: The artificial middle ear mucosa consisted of the single layer of epithelial cells, the basal membrane, and the underlying connective tissue. Electron microscopy revealed the presence of tight junctions and adherence junctions on the apical side, and adhesion complexes made of desmosomes. The reconstituted mucosa expressed genes of mucin, strongly suggesting that the artificial middle ear mucosa was capable of secreting mucus proteins. The DiI-labeled artificial middle ear mucosa implanted into the middle ear cavity was well engrafted and associated with host tissues.


Subject(s)
Artificial Organs , Ear, Middle/cytology , Guided Tissue Regeneration/methods , Mucous Membrane/cytology , Mucous Membrane/transplantation , Tissue Engineering/methods , Tissue Transplantation/methods , Tympanoplasty/methods , Animals , Cell Division/physiology , Collagen , Culture Media , Epithelial Cells/cytology , Epithelial Cells/transplantation , Fibroblasts/cytology , Fibroblasts/transplantation , Gene Expression Regulation , Keratins/ultrastructure , Mastoid/cytology , Mastoid/surgery , Microscopy, Electron , Mucins/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Vimentin/ultrastructure
16.
PLoS One ; 11(6): e0157451, 2016.
Article in English | MEDLINE | ID: mdl-27304995

ABSTRACT

Intermediate filament (IF) elongation proceeds via full-width "mini-filaments", referred to as "unit-length" filaments (ULFs), which instantaneously form by lateral association of extended coiled-coil complexes after assembly is initiated. In a comparatively much slower process, ULFs longitudinally interact end-to-end with other ULFs to form short filaments, which further anneal with ULFs and with each other to increasingly longer filaments. This assembly concept was derived from time-lapse electron and atomic force microscopy data. We previously have quantitatively verified this concept through the generation of time-dependent filament length-profiles and an analytical model that describes assembly kinetics well for about the first ten minutes. In this time frame, filaments are shorter than one persistence length, i.e. ~1 µm, and thus filaments were treated as stiff rods associating via their ends. However, when filaments grow several µm in length over hours, their flexibility becomes a significant factor for the kinetics of the longitudinal annealing process. Incorporating now additional filament length distributions that we have recorded after extended assembly times by total internal reflection fluorescence microscopy (TIRFM), we developed a Monte Carlo simulation procedure that accurately describes the underlying assembly kinetics for large time scales.


Subject(s)
Cytoplasm/metabolism , Desmin/metabolism , Intermediate Filaments/metabolism , Keratin-18/metabolism , Keratin-8/metabolism , Vimentin/metabolism , Algorithms , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Desmin/ultrastructure , Humans , Intermediate Filaments/ultrastructure , Keratin-18/ultrastructure , Keratin-8/ultrastructure , Kinetics , Microscopy, Atomic Force , Microscopy, Electron , Microscopy, Fluorescence , Monte Carlo Method , Time Factors , Time-Lapse Imaging/methods , Vimentin/ultrastructure
17.
Oncogene ; 16(26): 3423-34, 1998 Jul 02.
Article in English | MEDLINE | ID: mdl-9692550

ABSTRACT

The temperature-sensitive mutant tsp53val135 accumulates in the cytoplasm of cells kept at the non-permissive temperature (39 degrees C), but is rapidly transported into the cell nucleus at the permissive temperature (30 degrees C). tsp53 thus may serve as a model for analysing cellular parameters influencing the subcellular location of p53. Here we provide evidence that retention of tsp53 in the cytoplasm at the non-permissive temperature is due to cytoskeletal anchorage of the p53 protein. Two sublines of C6 rat glioma cells differing in their expression of the intermediate filament protein vimentin (vimentin expressing or vimentin negative cells) were stably transfected with a vector encoding tsp53. Whereas cells of vimentin expressing C6 subclones retained tsp53 in the cytoplasm at the non-permissive temperature, cells of vimentin negative subclones exclusively harbored the tsp53 within their nuclei. Intermediate filament deficient cells that had been reconstituted with a full length vimentin protein again showed a cytoplasmic localization of tsp53, whereas in cells expressing a C-terminally truncated (tail-less) vimentin tsp53 localized to the nucleus. We conclude that cytoplasmic sequestration of tsp53 requires an intact intermediate filament system.


Subject(s)
Cell Compartmentation , Cytoplasm/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Vimentin/metabolism , Actins/metabolism , Animals , Biological Transport , Cell Compartmentation/drug effects , Cell Nucleus/metabolism , Cytochalasin B/pharmacology , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Fluorescent Antibody Technique , Glioma , Mice , Mutation , Rats , Tubulin/metabolism , Tumor Cells, Cultured , Vimentin/ultrastructure , Vinblastine/pharmacology
18.
Biochim Biophys Acta ; 1702(1): 53-65, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15450850

ABSTRACT

Atomic force microscopy (AFM) was used to study the morphology of vimentin intermediate filaments (IFs) and their assembly intermediates. At each time after initiation of IF assembly in vitro of recombinant mouse vimentin, the sample was fixed with 0.1% glutaraldehyde and then applied to AFM analysis. When mature vimentin IFs were imaged in air on mica, they appeared to have a width of approximately 28 nm, a height of approximately 4 nm and a length of several micrometers. Taking into account the probe tip's distortion effect, the exact width was evaluated to be approximately 25 nm, suggesting that the filaments flatten on the substrate rather than be cylindrical with a diameter of approximately 10 nm. Vimentin IFs in air clearly demonstrated approximately 21-nm repeating patterns along the filament axis. The three-dimensional profiles of vimentin IFs indicated that the characteristic patterns were presented by repeating segments with a convex surface. The repeating patterns close to 21 nm were also observed by AFM analysis in a physiological solution condition, suggesting that the segments along the filaments are an intrinsic substructure of vimentin IFs. In the course of IF assembly, assembly intermediates were analyzed in air. Many short filaments with a full-width and an apparent length of approximately 78 nm (evaluated length approximately 69 nm) were observed immediately after initiation of the assembly reaction. Interestingly, the short full-width filaments appeared to be composed of the four segments. Further incubation enabled the short full-width filaments to anneal longitudinally into longer filaments with a distinct elongation step of approximately 40 nm, which corresponds to the length of the two segments. To explain these observations, we propose a vimentin IF formation model in which vimentin dimers are supercoiling around the filament axis.


Subject(s)
Intermediate Filaments/chemistry , Intermediate Filaments/ultrastructure , Microscopy, Atomic Force/methods , Vimentin/chemistry , Vimentin/ultrastructure , Animals , Fixatives , Glutaral , In Vitro Techniques , Mice , Recombinant Proteins/chemistry , Recombinant Proteins/ultrastructure
19.
J Mol Biol ; 340(1): 97-114, 2004 Jun 25.
Article in English | MEDLINE | ID: mdl-15184025

ABSTRACT

We have developed an assembly protocol for the intermediate filament (IF) protein vimentin based on a phosphate buffer system, which enables the dynamic formation of authentic IFs. The advantage of this physiological buffer is that analysis of the subunit interactions by chemical cross-linking of internal lysine residues becomes feasible. By this system, we have analyzed the potential interactions of the coiled-coil rod domains with one another, which are assumed to make a crucial contribution to IF formation and stability. We show that headless vimentin, which dimerizes under low salt conditions, associates into tetramers of the A(22)-type configuration under assembly conditions, indicating that one of the effects of increasing the ionic strength is to favor coil 2-coil 2 interactions. Furthermore, in order to obtain insight into the molecular interactions that occur during the first phase of assembly of full-length vimentin, we employed a temperature-sensitive variant of human vimentin, which is arrested at the "unit-length filament" (ULF) state at room temperature, but starts to elongate upon raising the temperature to 37 degrees C. Most importantly, we demonstrate by cross-linking analysis that ULF formation predominantly involves A(11)-type dimer-dimer interactions. The presence of A(22) and A(12) cross-linking products in mature IFs, however, indicates that major rearrangements do occur during the longitudinal annealing and radial compaction steps of IF assembly.


Subject(s)
Vimentin/chemistry , Buffers , Cross-Linking Reagents , Dimerization , Humans , Phosphates , Temperature , Ultracentrifugation , Vimentin/ultrastructure
20.
J Mol Biol ; 298(5): 817-32, 2000 May 19.
Article in English | MEDLINE | ID: mdl-10801351

ABSTRACT

Nearly all intermediate filament proteins exhibit a highly conserved amino acid motif (YRKLLEGEE) at the C-terminal end of their central alpha-helical rod domain. We have analyzed its contribution to the various stages of assembly by using truncated forms of Xenopus vimentin and mouse desmin, VimIAT and DesIAT, which terminate exactly before this motif, by comparing them with the wild-type and tailless proteins. It is surprising that in buffers of low ionic strength and high pH where the full-length proteins form tetramers, both VimIAT and DesIAT associated into various high molecular weight complexes. After initiation of assembly, both VimIAT and DesIAT aggregated into unit-length-type filaments, which rapidly longitudinally annealed to yield filaments of around 20 nm in diameter. Mass measurements by scanning transmission electron microscopy revealed that both VimIAT and DesIAT filaments contained considerably more subunits per cross-section than standard intermediate filaments. This indicated that the YRKLLEGEE-motif is crucial for the formation of authentic tetrameric complexes and also for the control of filament width, rather than elongation, during assembly. To determine the structure of the YRKLLEGEE domain, we grew crystals of peptides containing the last 28 amino acid residues of coil 2B, chimerically fused at its amino-terminal end to the 31 amino acid-long leucine zipper domain of the yeast transcription factor GCN4 to facilitate appropriate coiled-coil formation. The atomic structure shows that starting from Tyr400 the two helices gradually separate and that the coiled coil terminates with residue Glu405 while the downstream residues fold away from the coiled-coil axis.


Subject(s)
Consensus Sequence , Intermediate Filament Proteins/chemistry , Intermediate Filament Proteins/ultrastructure , Amino Acid Motifs , Amino Acid Sequence , Animals , Crystallography, X-Ray , Desmin/chemistry , Desmin/metabolism , Desmin/ultrastructure , Hydrogen-Ion Concentration , Intermediate Filament Proteins/metabolism , Mice , Microscopy, Electron, Scanning Transmission , Models, Molecular , Molecular Sequence Data , Molecular Weight , Osmolar Concentration , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Fragments/ultrastructure , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/ultrastructure , Sequence Alignment , Ultracentrifugation , Vimentin/chemistry , Vimentin/metabolism , Vimentin/ultrastructure , Viscosity , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL